TGF-β1 mediates hypoxia-preconditioned olfactory mucosa mesenchymal stem cells improved neural functional recovery in Parkinson's disease models and patients.

IF 16.7 2区 医学 Q1 MEDICINE, GENERAL & INTERNAL Military Medical Research Pub Date : 2024-07-22 DOI:10.1186/s40779-024-00550-7
Yi Zhuo, Wen-Shui Li, Wen Lu, Xuan Li, Li-Te Ge, Yan Huang, Qing-Tao Gao, Yu-Jia Deng, Xin-Chen Jiang, Zi-Wei Lan, Que Deng, Yong-Heng Chen, Yi Xiao, Shuo Lu, Feng Jiang, Zuo Liu, Li Hu, Yu Liu, Yu Ding, Zheng-Wen He, De-An Tan, Da Duan, Ming Lu
{"title":"TGF-β1 mediates hypoxia-preconditioned olfactory mucosa mesenchymal stem cells improved neural functional recovery in Parkinson's disease models and patients.","authors":"Yi Zhuo, Wen-Shui Li, Wen Lu, Xuan Li, Li-Te Ge, Yan Huang, Qing-Tao Gao, Yu-Jia Deng, Xin-Chen Jiang, Zi-Wei Lan, Que Deng, Yong-Heng Chen, Yi Xiao, Shuo Lu, Feng Jiang, Zuo Liu, Li Hu, Yu Liu, Yu Ding, Zheng-Wen He, De-An Tan, Da Duan, Ming Lu","doi":"10.1186/s40779-024-00550-7","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra (SN). Activation of the neuroinflammatory response has a pivotal role in PD. Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach for various nerve injuries, but there are limited reports on their use in PD and the underlying mechanisms remain unclear.</p><p><strong>Methods: </strong>We investigated the effects of clinical-grade hypoxia-preconditioned olfactory mucosa (hOM)-MSCs on neural functional recovery in both PD models and patients, as well as the preventive effects on mouse models of PD. To assess improvement in neuroinflammatory response and neural functional recovery induced by hOM-MSCs exposure, we employed single-cell RNA sequencing (scRNA-seq), assay for transposase accessible chromatin with high-throughput sequencing (ATAC-seq) combined with full-length transcriptome isoform-sequencing (ISO-seq), and functional assay. Furthermore, we present the findings from an initial cohort of patients enrolled in a phase I first-in-human clinical trial evaluating the safety and efficacy of intraspinal transplantation of hOM-MSC transplantation into severe PD patients.</p><p><strong>Results: </strong>A functional assay identified that transforming growth factor-β1 (TGF-β1), secreted from hOM-MSCs, played a critical role in modulating mitochondrial function recovery in dopaminergic neurons. This effect was achieved through improving microglia immune regulation and autophagy homeostasis in the SN, which are closely associated with neuroinflammatory responses. Mechanistically, exposure to hOM-MSCs led to an improvement in neuroinflammation and neural function recovery partially mediated by TGF-β1 via activation of the anaplastic lymphoma kinase/phosphatidylinositol-3-kinase/protein kinase B (ALK/PI3K/Akt) signaling pathway in microglia located in the SN of PD patients. Furthermore, intraspinal transplantation of hOM-MSCs improved the recovery of neurologic function and regulated the neuroinflammatory response without any adverse reactions observed in patients with PD.</p><p><strong>Conclusions: </strong>These findings provide compelling evidence for the involvement of TGF-β1 in mediating the beneficial effects of hOM-MSCs on neural functional recovery in PD. Treatment and prevention of hOM-MSCs could be a promising and effective neuroprotective strategy for PD. Additionally, TGF-β1 may be used alone or combined with hOM-MSCs therapy for treating PD.</p>","PeriodicalId":18581,"journal":{"name":"Military Medical Research","volume":"11 1","pages":"48"},"PeriodicalIF":16.7000,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265117/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Military Medical Research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s40779-024-00550-7","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, GENERAL & INTERNAL","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra (SN). Activation of the neuroinflammatory response has a pivotal role in PD. Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach for various nerve injuries, but there are limited reports on their use in PD and the underlying mechanisms remain unclear.

Methods: We investigated the effects of clinical-grade hypoxia-preconditioned olfactory mucosa (hOM)-MSCs on neural functional recovery in both PD models and patients, as well as the preventive effects on mouse models of PD. To assess improvement in neuroinflammatory response and neural functional recovery induced by hOM-MSCs exposure, we employed single-cell RNA sequencing (scRNA-seq), assay for transposase accessible chromatin with high-throughput sequencing (ATAC-seq) combined with full-length transcriptome isoform-sequencing (ISO-seq), and functional assay. Furthermore, we present the findings from an initial cohort of patients enrolled in a phase I first-in-human clinical trial evaluating the safety and efficacy of intraspinal transplantation of hOM-MSC transplantation into severe PD patients.

Results: A functional assay identified that transforming growth factor-β1 (TGF-β1), secreted from hOM-MSCs, played a critical role in modulating mitochondrial function recovery in dopaminergic neurons. This effect was achieved through improving microglia immune regulation and autophagy homeostasis in the SN, which are closely associated with neuroinflammatory responses. Mechanistically, exposure to hOM-MSCs led to an improvement in neuroinflammation and neural function recovery partially mediated by TGF-β1 via activation of the anaplastic lymphoma kinase/phosphatidylinositol-3-kinase/protein kinase B (ALK/PI3K/Akt) signaling pathway in microglia located in the SN of PD patients. Furthermore, intraspinal transplantation of hOM-MSCs improved the recovery of neurologic function and regulated the neuroinflammatory response without any adverse reactions observed in patients with PD.

Conclusions: These findings provide compelling evidence for the involvement of TGF-β1 in mediating the beneficial effects of hOM-MSCs on neural functional recovery in PD. Treatment and prevention of hOM-MSCs could be a promising and effective neuroprotective strategy for PD. Additionally, TGF-β1 may be used alone or combined with hOM-MSCs therapy for treating PD.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
TGF-β1介导缺氧预处理的嗅粘膜间充质干细胞改善帕金森病模型和患者的神经功能恢复。
背景:帕金森病(PD)是一种以黑质(SN)多巴胺能神经元变性为特征的神经退行性疾病。神经炎症反应的激活在帕金森病中起着关键作用。间充质干细胞(MSCs)已成为治疗各种神经损伤的一种有前途的方法,但将其用于帕金森病的报道有限,其潜在机制仍不清楚:我们研究了临床级低氧预处理嗅粘膜间充质干细胞(hOM)对帕金森病模型和患者神经功能恢复的影响,以及对帕金森病小鼠模型的预防作用。为了评估暴露于hOM-MSCs诱导的神经炎症反应改善和神经功能恢复,我们采用了单细胞RNA测序(scRNA-seq)、转座酶可访问染色质高通量测序(ATAC-seq)结合全长转录组同工酶测序(ISO-seq)和功能测试。此外,我们还介绍了一项I期首次人体临床试验的首批入组患者的研究结果,该试验评估了将hOM-间充质干细胞移植到重度帕金森病患者椎管内的安全性和有效性:功能检测发现,hOM-间充质干细胞分泌的转化生长因子-β1(TGF-β1)在调节多巴胺能神经元线粒体功能恢复方面发挥了关键作用。这种作用是通过改善与神经炎症反应密切相关的SN中的小胶质细胞免疫调节和自噬平衡来实现的。从机理上讲,暴露于hOM-间充质干细胞可导致神经炎症的改善和神经功能的恢复,而这部分是由TGF-β1通过激活位于帕金森病患者SN的小胶质细胞中的无性淋巴瘤激酶/磷脂酰肌醇-3-激酶/蛋白激酶B(ALK/PI3K/Akt)信号通路介导的。此外,hOM-间充质干细胞的椎管内移植改善了帕金森病患者神经功能的恢复并调节了神经炎症反应,且未观察到任何不良反应:这些发现为TGF-β1参与介导hOM-间充质干细胞对帕金森病神经功能恢复的有益影响提供了令人信服的证据。hOM-间充质干细胞的治疗和预防可能是治疗帕金森病的一种前景广阔且有效的神经保护策略。此外,TGF-β1可单独或与hOM-间充质干细胞疗法联合用于治疗帕金森病。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Military Medical Research
Military Medical Research Medicine-General Medicine
CiteScore
38.40
自引率
2.80%
发文量
485
审稿时长
8 weeks
期刊介绍: Military Medical Research is an open-access, peer-reviewed journal that aims to share the most up-to-date evidence and innovative discoveries in a wide range of fields, including basic and clinical sciences, translational research, precision medicine, emerging interdisciplinary subjects, and advanced technologies. Our primary focus is on modern military medicine; however, we also encourage submissions from other related areas. This includes, but is not limited to, basic medical research with the potential for translation into practice, as well as clinical research that could impact medical care both in times of warfare and during peacetime military operations.
期刊最新文献
Hans Chinese consume less O2 for muscular work than european-american. Exosome autoantibody biomarkers for detection of lung cancer. International Alliance of Urolithiasis (IAU) consensus on miniaturized percutaneous nephrolithotomy. Mechanism of lactic acidemia-promoted pulmonary endothelial cells death in sepsis: role for CIRP-ZBP1-PANoptosis pathway. Microenvironment-responsive nanomedicines: a promising direction for tissue regeneration.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1